CN112074264B - Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof - Google Patents

Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof Download PDF

Info

Publication number
CN112074264B
CN112074264B CN201980030336.3A CN201980030336A CN112074264B CN 112074264 B CN112074264 B CN 112074264B CN 201980030336 A CN201980030336 A CN 201980030336A CN 112074264 B CN112074264 B CN 112074264B
Authority
CN
China
Prior art keywords
target protein
growth factor
extracellular vesicles
extracellular vesicle
extracellular
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201980030336.3A
Other languages
Chinese (zh)
Other versions
CN112074264A (en
Inventor
尹智爱
郑现旭
秋珍镐
宋洙珉
宋芝闰
金容仁
赵胜纪
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Medy Tox Inc
Original Assignee
Medy Tox Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medy Tox Inc filed Critical Medy Tox Inc
Publication of CN112074264A publication Critical patent/CN112074264A/en
Application granted granted Critical
Publication of CN112074264B publication Critical patent/CN112074264B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/14Fungi; Culture media therefor
    • C12N1/16Yeasts; Culture media therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/02Cosmetics or similar toiletry preparations characterised by special physical form
    • A61K8/14Liposomes; Vesicles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/062Ascomycota
    • A61K36/064Saccharomycetales, e.g. baker's yeast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1808Epidermal growth factor [EGF] urogastrone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/97Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution from algae, fungi, lichens or plants; from derivatives thereof
    • A61K8/9728Fungi, e.g. yeasts
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/96Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution
    • A61K8/99Cosmetics or similar toiletry preparations characterised by the composition containing materials, or derivatives thereof of undetermined constitution from microorganisms other than algae or fungi, e.g. protozoa or bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/5005Wall or coating material
    • A61K9/5063Compounds of unknown constitution, e.g. material from plants or animals
    • A61K9/5068Cell membranes or bacterial membranes enclosing drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q17/00Barrier preparations; Preparations brought into direct contact with the skin for affording protection against external influences, e.g. sunlight, X-rays or other harmful rays, corrosive materials, bacteria or insect stings
    • A61Q17/04Topical preparations for affording protection against sunlight or other radiation; Topical sun tanning preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/004Aftersun preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/485Epidermal growth factor [EGF], i.e. urogastrone
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factor [FGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/65Insulin-like growth factors, i.e. somatomedins, e.g. IGF-1, IGF-2
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N1/00Microorganisms, e.g. protozoa; Compositions thereof; Processes of propagating, maintaining or preserving microorganisms or compositions thereof; Processes of preparing or isolating a composition containing a microorganism; Culture media therefor
    • C12N1/20Bacteria; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0051Oxidoreductases (1.) acting on a sulfur group of donors (1.8)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y108/00Oxidoreductases acting on sulfur groups as donors (1.8)
    • C12Y108/01Oxidoreductases acting on sulfur groups as donors (1.8) with NAD+ or NADP+ as acceptor (1.8.1)
    • C12Y108/01008Protein-disulfide reductase (1.8.1.8), i.e. thioredoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/41Particular ingredients further characterized by their size
    • A61K2800/412Microsized, i.e. having sizes between 0.1 and 100 microns
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/74Bacteria
    • A61K35/741Probiotics
    • A61K35/744Lactic acid bacteria, e.g. enterococci, pediococci, lactococci, streptococci or leuconostocs
    • A61K35/747Lactobacilli, e.g. L. acidophilus or L. brevis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/02Preparations for care of the skin for chemically bleaching or whitening the skin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Toxicology (AREA)
  • Dermatology (AREA)
  • Mycology (AREA)
  • Biotechnology (AREA)
  • Birds (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Immunology (AREA)
  • Botany (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Dispersion Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Gerontology & Geriatric Medicine (AREA)
  • Medical Informatics (AREA)

Abstract

The present invention relates to an extracellular vesicle (Extracellular vesicles) derived from a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins, an extracellular vesicle isolated from said recombinant microorganism, and uses thereof.

Description

Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof
Cross Reference to Related Applications
The present application claims priority and benefit from korean patent application No.10-2018-0052157 filed on date 05-04 of 2018, which is incorporated herein by reference for all purposes as if fully set forth herein.
Technical Field
One or more embodiments relate to an extracellular vesicle derived from a recombinant microorganism comprising a polynucleotide encoding a target protein, and uses thereof.
Background
Most animal cells secrete extracellular vesicles of various sizes and compositions that originate from the inside of the cell. Both prokaryotes and eukaryotes are known to secrete extracellular vesicles.
Extracellular vesicles are membrane structural vesicles having diameters as small as about 20nm and as large as about 5 μm. Extracellular vesicles have heterogeneity in their size and composition, and have a number of different species, exosomes (exosomes, about 30nm to about 100 nm), nuclear exosomes (ectosomes), microvesicles (microvesicles, about 100nm to about 1000 nm), microparticles (microparticles), outer membrane vesicles (outer membrane vesicles), and the like. The characteristics of extracellular vesicles are affected by the characteristics of the cell from which they originate.
Intracellular substances (e.g., deoxyribonucleic acid (DNA; deoxyribo Nucleic Acid), ribonucleic acid (RNA; ribonucleic Acid), proteins, etc.) may be naturally carried in extracellular vesicles and secreted extracellularly. The extracellular vesicles have the same components as the components of the biological membrane, have high biocompatibility (biocompability), and have a size of nano-scale, so that the substance delivery efficiency is excellent. Therefore, studies for delivering drugs using extracellular vesicles instead of existing delivery systems such as liposomes are currently being conducted. However, when the target protein is supported in the extracellular vesicles, the supporting efficiency of the target protein into the extracellular vesicles is low. Therefore, there is a need for a technique capable of stably supporting a target protein in an extracellular vesicle with excellent efficiency.
Disclosure of Invention
Technical problem
One or more embodiments include extracellular vesicles derived from a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins, wherein the microorganism is a lactic acid bacteria or a yeast.
One or more embodiments include extracellular vesicles isolated from the recombinant microorganism.
One or more embodiments include compositions for delivering one or more target proteins to an individual comprising, as an active ingredient, extracellular vesicles derived from the recombinant microorganism and a carrier.
One or more embodiments include a method of treating a disease in an individual comprising administering the composition to the individual.
One or more embodiments include a method of applying a cosmetic to an individual comprising applying the composition to an individual.
One or more embodiments include a method of producing an extracellular vesicle, comprising: culturing the microorganism to obtain a culture; and isolating extracellular vesicles from the culture.
Additional aspects will be set forth in part in the description which follows, and in part will be obvious from the description, or may be learned by practice of the presented embodiments.
Technical proposal
Reference will now be made in detail to the embodiments illustrated in the drawings, wherein like elements are designated by like reference numerals. In this regard, the present embodiments may have different forms and should not be construed as limited to the embodiments set forth herein. Accordingly, the embodiments are described below to illustrate aspects of the present disclosure by referring only to the drawings.
An embodiment of an aspect provides an Extracellular Vesicle (EV) derived from a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins, wherein the microorganism is a lactic acid bacterium or a yeast.
The target protein may be linked to a signal peptide. In other words, the target protein may be a fusion protein of a signal peptide and a target protein. The recombinant microorganism may load the target protein in an increased amount in the extracellular vesicles. In this case, the recombinant microorganism may have an increased extracellular vesicle (extracellular vesicle; EV) carrying capacity compared to a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins without a signal peptide. The extracellular vesicle carrying capacity refers to the degree to which a target protein is included in an Extracellular Vesicle (EV) or the degree to which the target protein is expressed in an Extracellular Vesicle (EV). The extracellular vesicle-carrying capacity may refer to the carrying capacity of the parent microorganism compared to a parent microorganism that does not comprise the polynucleotide encoding the target protein.
The signal peptide may consist of SEQ ID NO:4, or the nucleotide sequence of SEQ ID NOS:21 to 60, or a sequence comprising or similar to any one of the sequences.
For the recombinant microorganism, the lactic acid bacteria may be of a genus selected from the group consisting of Lactobacillus, lactococcus, and Bifidobacterium. The lactic acid bacteria may be lactobacillus paracasei (Lactobacillus paracasei), lactobacillus delbrueckii (Lactobacillus brevis), or lactobacillus plantarum (Lactobacillus plantarum).
For the recombinant microorganism, the yeast may belong to a genus selected from the group consisting of Saccharomyces (Saccharomyces), pichia (Pichia), and Hansenula (Hansenula). Saccharomyces may be Saccharomyces cerevisiae. Pichia can be Pichia pastoris and Hansenula can be Hansenula polymorpha (Hansenula polymorpha).
In the recombinant microorganism, the target protein may be a growth factor, a cytokine, an antibody, an enzyme, an inhibitory protein, or a fragment thereof. The growth factor may be a fibroblast growth factor. The target protein may be selected from the group consisting of fibroblast growth factor (fibroblast growth factor: FGF), epidermal growth factor (EPIDERMAL GROWTH FACTOR: EGF), hepatocyte growth factor (hepatocyte growth factor: HGF), insulin-like growth factor (insulin-like growth factor: IGF), placental growth factor (placenta growth factor: PGF), platelet-derived growth factor (plalet-derived growth factor: PDGF), transforming growth factor (transforming growth factor: TGF), vascular endothelial growth factor (vascular endothelial growth factor: VEGF), thioredoxin (thioredoxin: TRX), interleukin-1 (interleukin-1: IL-1), interleukin-10, interleukin-22, interleukin-13, and tumor necrosis factor (tumor necrosis factor: TNF). The target protein may be selected from, for example, IL-22, EGF, IGF1, FGF1 (hereinafter also referred to as fibroblast growth factor (acidic fibroblast growth factor: aFGF)), FGF2 (hereinafter also referred to as basic fibroblast growth factor (basic fibroblast growth factor: bFGF)), FGF7 (hereinafter also referred to as keratinocyte growth factor (keratinocyte growth factor: KGF)), TGFa, and TRX.
For the recombinant microorganism, the signal peptide may be a polypeptide consisting of SEQ ID NO:4, or a signal peptide encoded by the nucleotide sequence of SEQ ID NOS:21 to 60. The gene encoding the signal peptide may be ligated so that the signal peptide is ligated to the N terminal of the target protein. The signal peptide may be naturally occurring or heterologous to the protein. The target protein may be a heterologous protein (heterologous protein) to the microorganism. The recombinant microorganism may express the target protein. The target protein may be carried in EV in a state where the signal peptide is cleaved. The target protein may be supported on the membrane of the EV or inside the EV.
In the recombinant microorganism, the polynucleotide encoding the target protein may be expressible. The polynucleotide may be operably linked to a transcription control sequence. The transcriptional control sequence may be a promoter, operator, enhancer, or terminator. The polynucleotide may be operably linked to a translation control sequence. The translational control sequences may be ribosome binding site (ribosome binding site) or ribosome entry site sequences (ribosome ENTRY SITE sequence). The polynucleotide may be incorporated into the genome of the microorganism or may be present separately. The polynucleotide may be included in a vector. The vector may be an expression vector. The vector may be a plasmid or a viral vector.
Embodiments of another aspect provide extracellular vesicles isolated from the recombinant microorganism.
Embodiments of a further aspect provide a composition for delivering one or more target proteins to an individual comprising as active ingredients extracellular vesicles derived from the recombinant microorganism and a carrier.
In embodiments regarding the recombinant microorganism and the composition, the extracellular vesicles may be isolated from the culture medium of the microorganism. In other words, the extracellular vesicles may be secreted extracellularly. The extracellular vesicles may have an average diameter of about 20nm to about 500nm, for example, about 20nm to about 200nm, or about 100nm to about 200 nm. The extracellular vesicles may include the target protein. The target protein may be located on or within the membrane of the EV.
The extracellular vesicles may be isolated by any method capable of isolating the extracellular vesicles from the culture broth. For example, the extracellular vesicles can be separated by centrifugation, ultracentrifugation, filtration through a filter, ultrafiltration, gel filtration chromatography, ion exchange chromatography, precipitation, immunoprecipitation, preflow electrophoresis, capillary electrophoresis, or a combination thereof. The separation method may include washing for removing impurities, concentration, and the like. The extracellular vesicles may be produced by the method of isolating the extracellular vesicles hereinafter. The extracellular vesicles may be produced by ultrafiltration of the microbial culture broth using an ultra-fine filter having a cut-off value of 10kD or more, e.g., 50kD or more, 100kD or more, 300kD or more, or 500kD or more. The extracellular vesicles are precipitated by ultracentrifugation of the microbial culture broth at 100,000Xg or more. The isolation may be produced by the production method of extracellular vesicles according to the seventh embodiment hereinafter.
In embodiments regarding the composition, the carrier may be physiologically acceptable, e.g., may be pharmaceutically or cosmetically acceptable. The carrier may include saline, sterile water, ringer's solution, buffer, cyclodextrin, dextrose solution, maltodextrin solution, glycerol, ethanol, liposomes, or combinations thereof, which are commonly administered. And, the carrier may include an antioxidant, a diluent, a dispersant, a surfactant, a binding agent, a lubricant, or a combination thereof.
The composition may have a dosage form for oral or parenteral administration. The parenterally administered dosage form may be a topical administration dosage form. The topical administration dosage form may be a dosage form for administration to the skin or mucosa. The parenterally administered dosage form may be a solution, suspension, emulsion, external skin formulation, spray, or powder puff dosage form.
The composition may be administered to the individual by application to the skin, to the mucous membranes, or by nasal administration, etc.
The amount administered may be changed by the weight, age, sex, health status, diet, administration time, administration method, excretion rate, and severity of disease of the patient. Daily dosage refers to an amount of the active ingredient sufficient to treat a reduced disease state by being administered to an individual in need of treatment. The administration amount is about 0.01 mg/day to 1000 mg/day, or about 0.01 mg/day to about 500 mg/day, based on an adult individual weighing 70kg, and divided administration may be performed once to several times per day at predetermined time intervals.
The composition may be a cosmetic composition. The cosmetic composition may include ingredients commonly used in cosmetic compositions. The cosmetic composition may include conventional adjuvants and carriers such as antioxidants, stabilizers, solubilizers, vitamins, pigments, fragrances.
The cosmetic composition may be a solution, suspension, emulsion, dough, gel, cream, lotion, flour, oil, powder foundation, emulsion foundation, wax foundation, or spray. The cosmetic composition may have a dosage form such as a nourishing cream, astringent lotion, soothing lotion, skin lotion, essence, nourishing gel or massage cream.
The composition may be used to promote the growth of fibroblasts or keratinocytes or the synthesis of collagen in an individual. The composition may be used to prevent skin aging or to reduce wrinkles. In this case, the target protein may be a growth factor.
The composition may be for locally delivering the target protein to the individual. The composition may be for Transdermal (TRANSDERMALLY), intradermal (INTRADERMALLY), oral, or mucosal (transmucosally) as well as intra-mucosal delivery.
For the composition, the individual may be a mammal. The mammal may be a human, dog, cat, horse, or pig.
Embodiments of another aspect provide a method of treating a disease in an individual comprising administering the composition to the individual. The individual may be a mammal. The mammal may be a human, dog, cat, horse, or pig. The disease may be an inflammatory disease, a wound, allergic dermatitis, psoriasis, or acne.
Embodiments of another aspect provide a method of applying a cosmetic to an individual comprising applying the composition to an individual. The application may refer to application to the skin of an individual. The application may refer to application to aged skin or wrinkled skin. The cosmetic method may be for improving aged skin or wrinkled skin.
Embodiments of another aspect provide a method of producing an extracellular vesicle, comprising: obtaining a culture by culturing the recombinant microorganism described above; and isolating extracellular vesicles from the culture.
The culturing may be in a medium useful for the growth of the microorganism. The cultivation may be performed under conditions known to be suitable for lactic acid bacteria or yeasts, such as temperature and stirring conditions.
The step of isolating the extracellular vesicles from the culture may comprise any method of isolating extracellular vesicles from a culture.
The separating may include: centrifuging the culture to obtain a supernatant; filtering the supernatant; and separating the filtrate by ultracentrifugation to obtain a precipitate.
In the separation, centrifugal separation may be performed at about 1,000 to about 20,000 xg. In the filtering, the filtering may be filtering using an ultra-fine filter. The filtration may be performed by ultrafiltration of the supernatant using a cut-off value of 10kD or more, for example, 50kD or more, 100kD or more, 300kD or more, or 500kD or more. In the step of ultracentrifugation separating the filtrate to obtain a precipitate, the ultracentrifugation separation may be performed at 100,000Xg or more, for example, at about 100,000Xg to about 200,000Xg.
The method may further comprise suspending the precipitate.
It should be understood that the embodiments described herein are to be regarded as illustrative rather than restrictive. Descriptions of features or aspects in various embodiments should be typically considered as pertaining to other similar features or aspects of other embodiments.
Although one or more embodiments have been described herein with reference to the accompanying drawings, it will be apparent to those of ordinary skill in the art that many more modifications are possible without departing from the spirit of the disclosure.
Drawings
These and/or other aspects will be appreciated and understood based on the embodiments and figures described below.
FIG. 1 is an expression vector for expressing a target protein in a yeast cell;
FIG. 2 shows the degree of expression of target proteins in supernatants and extracellular vesicles (extracellular vesicle; EV) derived from Saccharomyces cerevisiae (S.cerevisiae) transformed with p416G-MF-hEGF1 (IGF 1, FGF2, TGF alpha, and TRX);
FIG. 3 shows the extent of expression of target proteins in supernatants and extracellular vesicles isolated from S.cerevisiae transformed with p416G-hFGF1, p416G-MF-hFGF1, p 416G-hTRX, and p 416G-MF-hTRX;
FIG. 4 shows the effect of yeast-derived extracellular vesicles containing growth factors on cell proliferation;
FIG. 5 shows IL-10 production in cells treated with extracellular vesicles derived from Saccharomyces cerevisiae (S.cerevisiae) including IL-22 or extracellular vesicles not including IL-22, respectively;
FIG. 6 shows the results of observing the degree of binding of extracellular vesicles derived from Saccharomyces cerevisiae (S. Cerevisiae) labeled with CFSE-labeled extracellular vesicles to cells by cell flow analysis;
FIG. 7 shows the results of measuring toxicity of yeast-derived extracellular vesicles to skin;
FIG. 8 shows the size and concentration distribution of extracellular vesicles derived from transformed lactic acid bacteria;
FIG. 9 shows the results of western blotting of extracellular vesicle solutions;
FIG. 10 shows the results of western blotting of extracellular vesicles derived from LMT1-21 transformed with recombinant pMT172, wherein the recombinant pMT172 comprises a gene encoding FGF1 fused or not fused to a signal peptide gene;
FIG. 11 shows the effect of growth factor-containing extracellular vesicles derived from lactic acid bacteria on cell proliferation;
FIG. 12 shows production of IL-10 in cells treated with extracellular vesicles derived from LMT1-21 that include IL-22 or that do not include IL-22 extracellular vesicles;
FIG. 13 shows the results of observing the extent of binding of CFSE-labeled extracellular vesicles to cells by cell flow analysis;
FIG. 14 shows the results of measuring toxicity of yeast-derived extracellular vesicles to skin;
FIG. 15 shows the results of observing the effect of growth factors or bare growth factors included in extracellular vesicles on epidermal cell proliferation or collagen formation;
fig. 16 shows the results of a stability test for EGF or bare EGF included in extracellular vesicles; and
Fig. 17 shows the results of stability test for FGF2 or naked FGF2 included in extracellular vesicles.
Best mode for carrying out the invention
Hereinafter, the present disclosure is described in further detail by way of examples. However, these examples are merely illustrative of the present disclosure and are not intended to limit the scope of the present disclosure.
Example 1: extracellular vesicles derived from yeast cells (Extracellular Vesicle; EV)
Recombinant yeast for expressing the target protein is prepared, and extracellular vesicles are isolated from the yeast. The specific process is as follows. Saccharomyces cerevisiae (Saccharomyces cerevisiae) was used as yeast cells.
1. Preparation of expression vectors
FIG. 1 shows an expression vector for expressing a target protein in a yeast cell. The expression vector was prepared using the sequence of plasmid pRS416 GPD (SEQ ID: NO. 1), the target proteins hEGF1, hIGF1, hFGF2, HTGF ALPHA, and hTRX. hEGF1, hFGF2, HTGF ALPHA, and hTRX have the amino acid sequence of SEQ ID: amino acid sequences of NOS 14, 15, 12, 13, 17, and 18, which are represented by SEQ ID NOS: nucleotide sequences of NOS 5, 6, 7, 8, 10 and 11. FGF7 can have the amino acid sequence of SEQ ID NO:9, the amino acid sequence of which may be a nucleotide sequence encoded by SEQ ID NO:9, and an amino acid sequence encoded by the nucleotide sequence of 9.
The vector in FIG. 1 was designated as p416G-MF-hEFG1 (IGF 1, FGF2, TGF alpha, and TRX) according to the target protein.
First, target protein genes optimized for codons, i.e., human EGF1, IGF1, FGF2, TGF alpha, and TRX genes, were synthesized by MicroGene according to the codon usage frequency of S.cerevisiae, as required. Each gene was made into the expression vector in FIG. 1 by using the p416GPD vector (ATCC 87360) (SEQ ID NO: 1). The expression vector of FIG. 1 includes a sequence of a polynucleotide (SEQ ID NO: 4) encoding the mating factor (mating factor) alpha-1 signal peptide (MF) of S.cerevisiae linked upstream of the target protein gene. As a control group, a gene to which a polynucleotide (SEQ ID NO: 4) encoding a signal peptide (MF) was not ligated was used. Also, a vector was prepared in the same manner using the p426GPD vector (ATCC 87361) (SEQ ID NO: 2) instead of the p416GPD (ATCC 87360) vector. The p416GPD vector is a vector which exists in a cell in a low copy (low copy), and the p426GPD vector exists in a cell in a high copy (high copy). In p416GPD and p426GPD, GPD refers to the nucleotide sequence of the promoter GPD (SEQ ID NO: 3).
In FIG. 1, the vector comprises the CEN/Ars sequence as the replication origin of S.cerevisiae, the ampicillin resistance gene (AMPICILLIN RESISTANCE GENE: ampr) sequence, the ColE1 ori sequence as the replication origin of E.coli (ESCHERICHIA COLI; E.coli), the promoter GPD sequence as the promoter sequence of S.cerevisiae, the SCCYCTERM sequence as the CYCterminator sequence of S.cerevisiae, the F1 ori sequence as the replication origin of phage, the promoter of S.cerevisiae, the ORF, terminator sequence (ScURA p-URA 3).
2. Expression of target proteins in yeast
The p416G-MF-hEGF1 (IGF 1, FGF2, TGFalpha, and TRX) vector was transformed into the S.cerevisiae CEN.PK2-1 strain according to the LiCl method. The resulting transformed strain was grown in 2mL of minimal ura-drop out medium (Yeast nitrogen base without amino acids) without an amino yeast nitrogen source (Sigma-Aldrich: cat. No. Y0626) 6.7 g/L), and YEAST SYNTHETIC drop-out without uracil (Sigma-Aldrich: cat.No. Y1501) 1.92g/L, glucose 2 (w/v)% were cultured once for one day, and the cultured strain was inoculated into 15ml of minimum ura-drop out medium including 1% acid casein (Casamino acids) so that the original OD 600 was 0.5, followed by the present culture. The culture was performed at 30℃for two days while stirring at 220rpm, and a sample group from which the supernatant of the cells had been removed was prepared and used as it is. And, the supernatant was filtered using a 100kDa cut-off membrane (Amicon Ultra-15 Centrifugal Filter Unit with Ultracel-10membrane (100K) millipore: cat. No. UFC 910024) to obtain a concentrated filtrate, and the filtrate was ultracentrifuged (ultracentrifugation) at 150,000Xg for 2 hours to separate Extracellular Vesicles (EV) and suspend it in 1ml of phosphate buffered saline (phosphate buffer saline; PBS). Here, the degree of protein expression was confirmed by performing western blotting (western blot) on the supernatant and the obtained extracellular vesicle sample.
FIG. 2 shows the extent of target protein expression in supernatants and extracellular vesicles isolated from Saccharomyces cerevisiae (S.cerevisiae) transformed with p416G-MF-hEGF1 (IGF 1, FGF2, TGF alpha, and TRX). Lanes 1 and 2 are western blot photographs showing the degree of expression of fusion proteins of each target protein linked to the signal peptide MF. Lane 1 includes all proteins expressed in yeast cells and isolated from the culture broth, i.e., all target proteins, either supported or not supported in extracellular vesicles. Lane 2 represents only the target protein loaded in extracellular vesicles. As shown in fig. 2, the target protein was carried in significantly increased amounts in extracellular vesicles in all six experimental groups.
FIG. 3 shows the extent of expression of target proteins in supernatants and extracellular vesicles isolated from S.cerevisiae transformed with p416G-hFGF1, p416G-MF-hFGF1, p416G-hTRX, and p 416G-MF-hTRX. In other words, fig. 3 shows the degree of capture by extracellular vesicles depending on the presence or absence of signal peptide.
Lane 1 shows the target protein within the extracellular vesicles obtained from the culture broth of the strain expressed without the signal peptide, and lane 2 shows the expression of the target protein within the extracellular vesicles obtained from the culture broth of the strain expressed and secreted together with the signal peptide sequence. As shown in fig. 3, the amount of target protein expressed in extracellular vesicles when expressed and secreted extracellularly, i.e., 2.648ng FGF1 and 35.518ng TRX in EV per billion, was significantly more compared to target protein expressed in cells, i.e., 0.667ng FGF1 and 0.047ng TRX in EV per billion.
3. Confirmation of the efficacy of growth factor-containing extracellular vesicles on cell proliferation
The concentration of each target protein within the extracellular vesicles isolated according to the method described in the second section was measured, and then each protein was diluted 10-fold with PBS and diluted in 4 steps starting from an initial concentration of 20 μl. mu.L of each dilution was added to a 96-well plate comprising NIH3T3 cell line or 5,000 HaCat cells in each well, followed by culturing at 37℃for 48 hours. Then, 10. Mu.L of CCK-8 kit (Cell Counting Kit-8 (Dojindo)) solution was added to each well. After two hours, the absorbance was measured at 450 nm. NIH3T3 cells were used for FGF1, FGF2, and IGF, and HaCat cells were used for TGFa and EGF.
FIG. 4 shows the effect of growth factor-containing extracellular vesicles isolated from yeast on cell proliferation. In FIG. 4, SC refers to Saccharomyces cerevisiae (Saccharomyces cerevisiae).
As a result, the extracellular vesicles comprising the target protein increase cell number dose-dependently. In FIG. 4, pichia EV-FGF1 and Hansenula EV-FGF1 were obtained by the same procedure as with S.cerevisiae, except that Pichia pastoris or Hansenula polymorpha (Hansenula polymorpha) transformed with FGF1 was used. In FIG. 4, the horizontal axis represents the concentration (w/v, ng/ml) of the target protein contained in the extracellular vesicles in the medium. The vertical axis represents the degree of cell proliferation in the used extracellular vesicle-containing solution compared with the control group (100%) and the results are represented by percentages.
4. Confirmation of IL-22 expression
The expression vector p426G-MF-IL-22 was made according to the description of the first section by using IL-22 as the target protein, and transformed into the S.cerevisiae CEN.PK2-1 strain according to the description of the second section. The same p426G-MF vector, excluding IL-22, was used as a control.
Specifically, colo205 cell lines were cultured in RPMI medium in 96-well plates and at 37℃for 48 hours, and then transformed with p426G-MF-IL22 vector and p426G-MF vector to purify extracellular vesicles derived from IL-22 expressing yeast or yeast not expressing IL-22, respectively. The extracellular vesicles were suspended in PBS at a concentration of 0.5mg/mL, added to 96-well plates at 20. Mu.L per well, and further incubated at 37℃for 6 hours. The expression level of IL-22 was then compared indirectly by the expression level of IL-10 by extracting the protein from the cell line. IL-22 has the sequence of SEQ ID: amino acid sequence of NO 19. IL-22 is known to promote IL-10 production.
FIG. 5 shows the IL-10 protein production confirmed using protein extracted from Colo205 cell lines treated with extracellular vesicles including IL-22 or extracellular vesicles not including IL-22, respectively. As shown in FIG. 5, in the case of culturing by contacting the extracellular vesicles including IL-22 with cells, the production amount of IL-10 protein was significantly increased, compared to the extracellular vesicles not including IL-22. In FIG. 5, lane 1 shows the degree of production of IL-10 protein by Colo205 cell lines treated with extracellular vesicles not including IL-22, and lane 2 shows the degree of production of IL-10 protein by Colo205 cell lines treated with extracellular vesicles including IL-22.
5. Fusion of yeast-derived extracellular vesicles and cells
Extracellular vesicles were isolated from the untransformed S.cerevisiae CEN.PK2-1 strain as described above. 1ml of the isolated extracellular vesicles (0.5 mg/ml PBS) were placed in a 5. Mu.M solution of CFSE (5-Carboxyfluorescein N-hydroxysuccinimidyl ester) at room temperature for 30 minutes. Next, residual CFSE was removed from the solution using PD-10 desalting column (DESALTING COLUMN) (GE) to obtain CFSE-labeled extracellular vesicles. After culturing NIH3T3 cell lines in RPMI medium 0.2mL in each well of 96-well plates at 37 ℃ for 48 hours, 10 μl (red) or 20 μl (green) of CFSE-labeled extracellular vesicles in PBS were added and further cultured at 37 ℃ for 24 hours. Thereafter, the cells were washed with PBS. The residual cells were passed through a flow cytometer (flow cytometer) and their fluorescence levels were measured. As a control group, 0.5. Mu.g/ml BSA was labeled with the same CFSE and 20. Mu.l of the product was used.
Fig. 6 shows the results of observing the extent of fusion of CFSE-labeled extracellular vesicles with cells by cell flow analysis. In fig. 6, the control (left graph) and experimental (right graph) groups show the results observed after CFSE-labeled extracellular vesicles were contacted with 10 μl (red) and 20 μl (green) of cells. As a result, as represented in the right graph of fig. 6, the cells were stained with CFSE, thereby confirming that extracellular vesicles are fused with the cells and that components causing extracellular vesicles are introduced into the cells. NIH3T3 cells are a standard fibroblast (fibroblastist) cell line.
6. Confirmation of toxicity of Yeast-derived extracellular vesicles to skin
Toxicity of yeast-derived extracellular vesicles to skin was measured by toxicity test on artificial skin according to OECD guidelines. Neoderm TM -ED (product of Taigo Science company) was used as artificial skin.
Extracellular vesicles derived from Saccharomyces cerevisiae, pichia pastoris, or Hansenula polymorpha (Hansenula polymorpha) were isolated. Extracellular vesicles derived from Saccharomyces cerevisiae (S.cerevisiae) were isolated according to the description in the second section. In addition to the use of Pichia pastoris (Pichia pastoris) and Hansenula polymorpha (Hansenula polymorpha), extracellular vesicles derived from Pichia pastoris (Pichia pastoris) or Hansenula polymorpha (Hansenula polymorpha) were isolated according to the description in the second section.
The isolated extracellular vesicles, PBS as a negative control group, and 5% SDS as a positive control group were coated on Neoderm TM -ED artificial skin in an amount of 30. Mu.L, respectively, and incubated at 37℃for 15 minutes. The artificial skin was then washed with PBS and precipitated in 2ml of assay medium (Taigo Science company) in a 12-well plate and incubated at 37 ℃ for an additional 42 hours.
The incubated artificial skin was scooped up and moved to a 0.3% MTT solution (0.3 mg/ml) and incubated at 37℃for three hours. Then, the artificial skin was again fished and each tissue was separated using an 8mm biopsy punch (biopsy punch), and the tissue was put into 500 μl of 0.04N HCL-isopropanol and decolorized for four hours. After absorbance was measured at 570nm, it was compared with absorbance in a control group to calculate survival (%). As a result, no toxicity was determined when the measured survival rate was an intermediate value or higher of the values of the positive control group and the negative control group. Survival was calculated according to the following equation.
Survival = test substance absorbance/negative control absorbance x 100
Figure 7 shows the measurement of toxicity of yeast derived extracellular vesicles to skin. In fig. 7, 1 represents a negative control group (PBS); 2 represents a positive control group (5% SDS); 3 represents an extracellular vesicle derived from s.cerevisiae; 4 represents extracellular vesicles derived from p. 5 represents extracellular vesicles derived from H.polymorpha.
Example 2: extracellular vesicles derived from lactic acid bacteria (LACTIC ACID bacteria: LAB) cells
Recombinant lactic acid bacteria expressing the target protein are prepared, and extracellular vesicles are isolated from the lactic acid bacteria. The specific process is as follows. Lactobacillus paracasei (Lactobacillus paracasei) LMT1-21 (KCTC 13422 BP), lactobacillus delbrueckii (Lactobacillus brevis) LMT1-46 (KCTC 13423 BP) and/or Lactobacillus plantarum (Lactobacillus plantarum) LMT1-9 (KCTC 13421 BP) were used as lactic acid bacteria cells.
1. Preparation of Gene expression vectors
For the target gene, a nucleotide sequence of a codon optimized for the lactic acid bacterium used was obtained from the amino acid sequence of the protein using a codon optimization tool (http:// sq. Idtdna. Com/CodonOpt), a sequence having recognition sequences for BamHI and XhoI restriction enzymes at both ends of the sequence was designed, and a DNA having the sequence was synthesized (Macrogen company, korea). BamHI and Xhol restriction enzymes were used to cleave the synthesized gene. Also, the parent vector pMT182-PR4 (SEQ ID NO: 20) was cut using the same restriction enzyme, purified using a gel purification kit (Gel purification kit), and dephosphorized using alkaline phosphatase (AP; alkaline phosphatase). The parent vector includes a promoter PR4 for expressing the target protein and a signal peptide SP4 (SEQ ID NO: 21) for secreting the target protein to the outside of the cell.
1. Mu.L of the vector DNA prepared by this method, 3. Mu.L of insert DNA, 0.5. Mu.L of T4DNA ligase (Takara Co., ltd., japan), and 1. Mu.L of buffer solution were added to 5.5. Mu.L of distilled water to make the total volume 10. Mu.L. The reaction solution was incubated at 16℃for 12 hours to carry out ligation, and the ligation product obtained was transformed into E.coli strain ten by the method of Sambrook et al (Sambrook et al Molecular Cloning: A laboratory manual,2nd ed.1989). The sequences of plasmids obtained from each group were analyzed and confirmed. FGF1, FGF2, EGF, IGF, KGF, TGFa, TRX and IL-22 were used as target proteins. Which have the respective SEQ ID: amino acid sequences of NOS1, 2,3, 4,5, 6, 7, and 8.
2. Lactic acid bacteria conversion
The cloned DNA obtained was transformed into three lactic acid bacteria. Each strain was cultured in 50mL of MRS until OD 600 was 0.5, centrifuged at 7,000rpm for 10 minutes at 4℃and washed twice with 25mL of ice-cold (ice-cold) EPS (containing 1mM K 2HPO4KH2PO4, pH7.4,1mM MgCl 2 and 0.5M sucrose). The reaction potential cells (competent cell) useful for electroporation (electroporation) were made by suspending the cells in 1mL of ice-cold EPS and stored in a freezer (deep freezer) at-80 ℃. mu.L of reaction potential cells and 1. Mu.g/. Mu.L of vector DNA were transferred to a tube and placed on ice for five minutes. Immediately after providing the pulse at 25. Mu.F, 8kV/cm, and 400ohms, 1mL of MRS broth was added and incubated at 37℃for about one hour. The cells were smeared on MRS medium including chloramphenicol at 10. Mu.g/ml and cultured at 37℃for 49 hours to obtain transformed cells.
3. Isolation of extracellular vesicles
From the transformed lactic acid bacteria (LACTIC ACID bacteria; LAB) strain obtained, KCTC13422BP strain was left to stand for 16 hours at 37℃in MRS liquid medium, and then it was treated with 2%
The (w/v) volume was inoculated into MRS liquid medium and left to stand for 16 hours. The culture obtained was centrifuged at 5,000Xg for 15 minutes to obtain a supernatant from which lactic acid bacteria were removed, and then the supernatant was ultrafiltered and concentrated 20 times by using a 100kDa cut-off (molecular weight cut-off; MWCO) ultrafiltration membrane. The concentrate was ultracentrifuged at 150,000Xg for 3 hours to obtain a pellet which was precipitated, and the pellet was resuspended with PBS to obtain an extracellular vesicle solution. The size and number of extracellular vesicles obtained were measured by using Nanosight NS300 (malvern). The results are shown in fig. 8.
Figure 8 shows the size and concentration profile of extracellular vesicles isolated from transformed lactic acid bacteria. In fig. 8, the horizontal axis represents diameter, and the vertical axis represents concentration (particles/ml). In FIG. 8, the lactic acid bacterium used was KCTC13422BP strain, and the target protein was FGF1.
As shown in fig. 8, 90% of the particles in the extracellular vesicles were distributed with a particle size of 80nm to 250 nm.
4. Confirmation of the Presence of target proteins in extracellular vesicles
Western blotting was performed on the EV solution obtained in the third section to confirm whether or not it was present in the target protein in EV. The extracellular vesicles were isolated from a KCTC13422BP strain (hereinafter also referred to as LMT 1-21) transformed with a vector obtained by cloning pMT182-PR4 using a gene encoding FGF1 or TRX. In this case, the gene is fused to a signal peptide, that is, SP4 sequence, or a sequence not fused thereto.
Western blotting was performed by the following method. 4x loading buffer (thermo), 10x reducing agent (thermo) was added to 5. Mu.L EV solution, and then electrophoresis was performed on a gel for sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE; sodium dodecyl sulfate-polyacrylamide gel electrophoresis). The proteins of the gel were transferred to nitrocellulose membranes, which were then blocked by incubation for two hours in physiological saline buffer (TBST; tris-buffered SALINE WITH TWEEN) containing Tween 20 as blocking solution, comprising 5% skim milk. Then, the membrane and the primary antibody were added to the blocking solution and incubated for two hours to induce antigen-antibody binding. After TBST washing, secondary antibodies were added. After one hour of placement, the components and location of the target proteins were confirmed by using an enhanced electrochemical (ECL; enhanced electrochemical) system.
Fig. 9 shows the results of western blotting of EV solutions. As shown in fig. 9, FGF1 and TRX were expressed in extracellular vesicles isolated from LMT1-21 transformed with a vector obtained by cloning pMT182-PR4 using FGF1 and TRX fused to a signal peptide, respectively, and thus, it was seen that FGF1 and TRX were present in extracellular vesicles.
FIG. 10 shows the results of protein immunoblotting of extracellular vesicles isolated from LMT1-21 transformed by using a recombinant vector comprising a gene encoding FGF1 fused with or not fused with a signal peptide gene, i.e., pMT182-PR4-FGF1 or pMT182-PR4-SP4-FGF1 vector. In fig. 10, lane 1 represents an extracellular vesicle in the case of expressing the FGF1 gene without using a signal peptide, and lane 2 represents an extracellular vesicle in the case of expressing a gene encoding a fusion protein of a signal peptide and FGF 1.
5. Confirmation of the efficacy of growth factor-containing extracellular vesicles derived from lactic acid bacteria on cell proliferation
According to the method described in the third section, extracellular vesicles isolated from 1L of lactic acid bacteria culture broth were suspended in 1ml of PBS. NIH3T3 cell lines (or HaCat cells) in DMEM medium were seeded (seeding) at 5,000 cells/well in each well of a 96-well plate and cultured at 37 ℃ for 48 hours. Then, 20. Mu.L of the solution containing growth factor-expressing extracellular vesicles or PBS as a control group was added to each well. After 48 hours of incubation under the same conditions, 10uL of CCK-8 kit (Dojindo) solution was added to each well. After two hours, the absorbance was measured at 450 nm. NIH3T3 cells were used for FGF1, FGF2, and IGF, and HaCat cells were used for KGF, TGFa, and EGF.
FIG. 11 shows the effect of growth factor-containing extracellular vesicles isolated from lactic acid bacteria on cell proliferation. In FIG. 11, LAB refers to lactic acid bacteria (LACTIC ACID bacteria) (KCTC 13422BP strain). As a result, the extracellular vesicles containing the target protein increase the cell concentration dose-dependently. In FIG. 11, the horizontal axis represents the concentration (w/v) of the target protein included in the extracellular vesicles. The vertical axis represents the degree of cell proliferation in the used extracellular vesicle-containing solution compared with the control group and the results are expressed as percentages.
6. Potency of extracellular vesicles including growth factors: confirmation of IL-10 expression
A vector expressing IL-22 was prepared according to the first and second sections and transformed into LMT 1-21. According to the third section, EV is isolated from LMT1-21 transformed with a vector expressing IL-22. To confirm whether the EV promotes the expression of IL-10 in cells, the presence of IL-22 was indirectly presumed.
Specifically, colo205 cell line was cultured in RPMI medium on a 96-well plate at 37℃for 48 hours, from which extracellular vesicles derived from either IL-22 expressing lactic acid bacteria or lactic acid bacteria not expressing IL-22 were isolated, suspended in PBS at a concentration of 0.5mg/mL, added at 20. Mu.L to each well and further cultured at 37℃for 6 hours. Then, a lysate (lysate) was obtained after extracting a protein from the cells, that is, cell lysis (lysis), and the expression level of IL-10 in the protein was compared.
FIG. 12 is a photograph of Western blotting of cell-derived proteins contacted with EV derived from LMT 1-21. In FIG. 12, lane 1 represents PBS, lane 2 represents EV derived from LMT1-21, and lane 3 represents EV derived from LMT1-21 expressing IL-22.
7. Fusion of extracellular vesicles and cells derived from lactic acid bacteria
From the untransformed lactic acid bacterial strain (KCTC 13422 BP), extracellular vesicles were isolated as described above. 1ml of the isolated extracellular vesicles (0.5 mg/ml) were left in a5. Mu.M CFSE solution at room temperature for 30 minutes. Next, residual CFSE was removed from the solution using PD-10 desalting column (DESALTING COLUMN) (GE) to obtain CFSE-labeled extracellular vesicles. After culturing NIH3T3 cell lines in 0.2mL RPMI medium for 48 hours in each well of 96-well plates, CFSE-labeled 10 μl (red) or 20uL (green) extracellular vesicles in PBS were added to each well and further cultured for 24 hours. Thereafter, the cells were washed with PBS. The residual cells were passed through a flow cytometer (flow cytometer) and their fluorescence levels were measured. As a control group, 0.5. Mu.g/ml BSA was labeled with the same CFSE and 20. Mu.l of the product was used.
Fig. 13 shows the results of observing the degree of fusion of CFSE-labeled extracellular vesicles with cells by cell flow analysis. In fig. 13, a control group (left graph) shows the results observed after CFSE-labeled BSA and cells were brought into contact with each other, and an experimental group (right graph) shows the results observed after CFSE-labeled 10 μl (red) of extracellular vesicles, 20 μl of extracellular vesicles (green) and cells were brought into contact with each other. As a result, as shown in the right graph of fig. 13, cells were stained with CFSE, whereby it was confirmed that extracellular vesicles were fused with cells and components of extracellular vesicles were introduced into cells. NIH3T3 cells are a standard fibroblast (fibroblastist) cell line.
8. Confirmation of toxicity of extracellular vesicles derived from lactic acid bacteria to skin
Toxicity of extracellular vesicles derived from lactic acid bacteria to skin was measured by toxicity test on artificial skin according to OECD guidelines. Neoderm TM -ED (product of Taigo Science company) was used as artificial skin.
Extracellular vesicles derived from LMT1-21, LMT1-9, or LMT1-46 will be isolated. These extracellular vesicles were isolated according to the description in the second section. The isolated extracellular vesicles, PBS as a control group, and 5% SDS as a positive control group were smeared on Neoderm TM -ED artificial skin in a fraction of 30. Mu.L each, followed by incubation at 37℃for 15 minutes. The artificial skin was then washed with PBS and immersed in 2ml of assay medium (Taigo Sicence company product) in a 12-well plate and incubated at 37 ℃ for a further 42 hours.
The incubated artificial skin was scooped up and moved into a 0.3% MTT solution (0.3 mg/ml) for incubation for 3 hours. Then, the artificial skin was again fished and tissue was separated using an 8mm biopsy punch (biopsy punch) and thrown into 500 μl of 0.04N HCl-isopropanol and decolorized for 4 hours. The absorbance at 570nm was measured and compared with the absorbance of the control group to calculate the survival (%). As a result, no toxicity was determined when the measured survival rate was an intermediate value or higher of the values of the positive control group and the negative control group. Survival was calculated according to the following equation.
Survival = test substance absorbance/negative control absorbance x 100
Figure 14 shows the measurement of the toxicity of extracellular vesicles derived from lactic acid bacteria to the skin. In FIG. 14, 1 represents a negative control group (PBS), 2 represents a positive control group (5% SDS), 3 represents extracellular vesicles derived from LMT1-46, 4 represents extracellular vesicles derived from LMT1-9, and 5 represents extracellular vesicles derived from LMT 1-21.
Example 3: comparison of cell proliferation efficiency of extracellular vesicles containing growth factors and cell proliferation efficiency of naked growth factors
1. Preparation of growth factors including extracellular vesicles
Extracellular vesicles comprising growth factors were isolated from Pichia pastoris transformed with p416G-MF-EGF, p416G-MF-FGF1 and p416G-MF-FGF2, respectively, in the same manner as in the third item of example 1. Each of them was suspended in PBS to adjust the concentration of EGF to 10ug/ml and the concentration of FGF1 or FGF2 to 1ug/ml. As a control group, bare EGF, FGF1, and FGF2 protein were purchased from AbCam and suspended in PBS until reaching the concentrations described above.
2. Comparison of the effects of cell proliferation of extracellular vesicles comprising growth factors and cell proliferation of naked growth factors
Artificial skin, neoderm TM -ED, was purchased from TaiGo Science company. The artificial skin was washed with PBS and 2mL of PBS, 2mL of the solution prepared as described above including extracellular vesicle growth factor, and 2mL of the control group solution prepared as described above including bare EGF, FGF1, or FGF2 protein were added to the wells of the 12-well plate, followed by further incubation at 37 ℃ for 24 hours. After three washes with PBS, the artificial skin was fixed in 4% paraformaldehyde solution (Sigma, usa) for 18 hours at 37 ℃ and frozen sections were made with Leica Biosystems. Immunohistochemistry (Immunohistochemistry; IHC) was performed by using an anti-Ki-67 antibody for EGF-EV and a control group and using an anti-collagen antibody for FGF1-EV and FGF2-EV (AbCam) and a control group, followed by the addition of 3,3'diaminobenzidine (3, 3' diaminobenzidine; DAB). The results were photographed under a microscope. Overall, the rich Ki-67 or collagen was observed as brown. Ki-67 is a biomarker for epidermal cell proliferation.
As shown in fig. 15, more favorable epidermal cell proliferation was observed from EGF-EV treatment, compared to treatment with the PBS and the control group (line a). And, better collagen synthesis was observed with FGF1-EV and FGF2-EV compared to PBS or control proteins (rows B and C). According to these results, the growth factors included in the extracellular vesicles are more effective for cell proliferation than the naked growth factors, regardless of the type of growth factor. And FGF2-EV is most effective compared to any other growth factor that is included in or not included in the extracellular vesicles.
Example 4: comparison of growth factor stability
1. EGF-EV stability compared to bare EGF stability
EGF-EV derived from Pichia pastoris and the control protein, that is, EGF protein not included in EV, were prepared by the same method as in the second item of example 1. Briefly, the EGF-EV or the EGF protein was suspended in 1ml of PBS until its concentration was 10ug/ml, and cultured at 40℃for 8 weeks. Once a week, for cell proliferation activity analysis, the samples were aliquoted and diluted with PBS to a concentration of 100 ng/ml.
HaCat cells in DMEM medium were seeded at a density of 5,000 cells/well in each well of a 96-well plate and cultured at 37 ℃ for 48 hours. Then, 20. Mu.L of EGF-EV, control protein, various samples of PBS were added thereto. The cells were cultured under the same conditions for 48 hours, and then 10uL of cell counting kit-8 (Dojindo) solution was added to each well. After two hours, the absorbance was measured at 450 nm.
As shown in fig. 16, EGF included in EV is more stable than bare EGF.
2. Stability of FGF2-EV compared to stability of naked FGF2
EGF2-EV derived from Pichia pastoris and the control protein, namely FGF-2 protein not included in EV, were prepared by the same method as in the second item of example 1. Briefly, the FGF2-EV or the FGF2 protein is suspended in 1ml of PBS until its concentration is 10ug/ml, and incubated for 4 weeks at ambient temperature. For cell proliferation activity analysis, each sample was periodically aliquoted and diluted with PBS to a concentration of 100 ng/ml.
NIH3T3 cells in DMEM medium were seeded at a density of 5,000 cells/well in each well of a 96-well plate and cultured at 37 ℃ for 48 hours. Then, 20uL of FGF2-EV, control protein, and various samples of PBS were added thereto. The cells were cultured under the same conditions for 48 hours, and then 10. Mu.L of cell counting kit-8 (Dojindo) solution was added to each well. After two hours, the absorbance was measured at 450 nm.
As shown in fig. 17, FGF2 included in EV was more stable than naked FGF 2.
Industrial applicability
Recombinant microorganisms according to an embodiment may be used to efficiently isolate extracellular vesicles or target proteins from the extracellular vesicles.
According to another embodiment, a composition for delivering the extracellular vesicles and target proteins to an individual may be used to efficiently deliver the target proteins to an individual.
According to another embodiment, a method of treating a disease in an individual may be used to effectively treat the disease.
According to another embodiment, a method of applying cosmetics to an individual may be used to effectively apply cosmetics to an individual.
According to another embodiment, a method of generating extracellular vesicles may be used to efficiently generate EV.

Claims (11)

1. An extracellular vesicle derived from a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins, wherein the microorganism is a yeast, wherein the yeast belongs to a genus selected from the group consisting of saccharomyces, pichia and hansenula, and wherein the target protein is linked to a signal peptide, whereby the microorganism carries the target protein in the extracellular vesicle in an increased amount compared to the target protein not linked to the signal peptide, wherein the signal peptide consists of SEQ ID NO:4, and the extracellular vesicles comprise the target protein,
Wherein the target protein is selected from one or more of the group consisting of fibroblast growth factor 1 (FGF 1), fibroblast growth factor 2 (FGF 2), insulin-like growth factor 1 (IGF 1), KGF, tgfα, TRX, and IL-22.
2. The extracellular vesicle of claim 1, wherein the extracellular vesicle is isolated from a culture broth of the recombinant microorganism.
3. The extracellular vesicle of claim 2, wherein the extracellular vesicle is isolated by precipitation of the microbial culture broth by ultracentrifugation at 100,000x g or more.
4. The extracellular vesicle of claim 1, wherein the extracellular vesicle has a diameter of 20nm to 500 nm.
5. An extracellular vesicle derived from a recombinant microorganism comprising one or more polynucleotides encoding one or more target proteins, the extracellular vesicle having a pharmaceutical use, wherein the microorganism is a yeast, wherein the yeast belongs to a genus selected from the group consisting of saccharomyces, pichia and hansenula, and wherein the target protein is linked to a signal peptide, whereby the microorganism carries the target protein in the extracellular vesicle in an increased amount compared to when the target protein is not linked to the signal peptide, wherein the signal peptide consists of SEQ ID NO:4, and the extracellular vesicle comprises the target protein, wherein the target protein is Epidermal Growth Factor (EGF).
6. A composition for delivering a target protein to an individual, comprising an extracellular vesicle according to any one of claims 1 to 5 as an active ingredient and a carrier, wherein the individual is a mammal, wherein the composition is for reducing wrinkles of skin, whitening skin, blocking ultraviolet light or reducing inflammation, wherein the target protein is selected from one or more of the group consisting of fibroblast growth factor 1 (FGF 1), fibroblast growth factor 2 (FGF 2), insulin-like growth factor 1 (IGF 1), KGF, tgfα, TRX and IL-22.
7. The composition of claim 6 for transdermal, intradermal, oral, transmucosal, or intramucosal delivery of a target protein.
8. The composition of claim 7, having pharmaceutical or cosmetic use.
9. A composition for delivering a target protein to an individual, the composition having a pharmaceutical use comprising an extracellular vesicle according to any one of claims 1 to 5 as an active ingredient and a carrier, wherein the individual is a mammal, wherein the composition is for reducing wrinkles in skin, whitening skin, blocking uv light or reducing inflammation, wherein the target protein is Epidermal Growth Factor (EGF).
10. Use of an extracellular vesicle according to any one of claims 1 to 5 in the manufacture of a medicament for reducing wrinkles in skin, whitening skin, blocking uv light or reducing inflammation, wherein the target protein is selected from one or more of the group consisting of fibroblast growth factor 1 (FGF 1), fibroblast growth factor 2 (FGF 2), epidermal Growth Factor (EGF), insulin-like growth factor 1 (IGF 1), KGF, tgfα, TRX and IL-22.
11. Use of the extracellular vesicles according to any one of claims 1 to 4 for the preparation of a cosmetic for reducing wrinkles in skin, whitening skin, blocking uv light or reducing inflammation, wherein the target protein is selected from one or more of the group consisting of fibroblast growth factor 1 (FGF 1), fibroblast growth factor 2 (FGF 2), insulin-like growth factor 1 (IGF 1), KGF, tgfα, TRX and IL-22.
CN201980030336.3A 2018-05-04 2019-05-03 Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof Active CN112074264B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR10-2018-0052157 2018-05-04
KR1020180052157A KR102015116B1 (en) 2018-05-04 2018-05-04 Extracellular vesicle isolated from recombinant microorganism comprising polynucleotide encoding target protein and use thereof
PCT/KR2019/005334 WO2019212293A1 (en) 2018-05-04 2019-05-03 Extracellular vesicles derived from recombinant microorganism including polynucleotide encoding target protein and use thereof

Publications (2)

Publication Number Publication Date
CN112074264A CN112074264A (en) 2020-12-11
CN112074264B true CN112074264B (en) 2024-05-07

Family

ID=68386667

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201980030336.3A Active CN112074264B (en) 2018-05-04 2019-05-03 Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof

Country Status (6)

Country Link
US (1) US20210169802A1 (en)
EP (1) EP3773423A4 (en)
JP (2) JP7297872B2 (en)
KR (1) KR102015116B1 (en)
CN (1) CN112074264B (en)
WO (1) WO2019212293A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2021521141A (en) * 2018-04-09 2021-08-26 オージェネシス インコーポレイテッド Bioxome particles, redoxomes, methods of their preparation, and compositions containing them.
US20220133826A1 (en) * 2019-02-01 2022-05-05 Da Vinci Universale Co., Ltd. Novel component for controlling biological function
US20230084480A1 (en) * 2020-01-20 2023-03-16 Ev Cell Biotech (Guangzhou) Co., Ltd. Extracellular vesicle and use thereof in skin products
KR102440312B1 (en) * 2020-08-28 2022-09-05 한국해양과학기술원 Thermally stable fgf7 polypeptide and use of the same
KR102633132B1 (en) * 2021-05-21 2024-02-05 (주)메디톡스 Composition Comprising Extracellular Vesicles Derived from Yeast, and methods related thereto
KR20230008997A (en) * 2021-07-08 2023-01-17 주식회사 래디안 Cosmetic composition comprising extracellular vesicles and lysate derived from yeast

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5821079A (en) * 1985-05-02 1998-10-13 Transgene S.A. Vectors for the expression and secretion of hirudin by transformed yeasts
KR19990064308A (en) * 1995-10-20 1999-07-26 알. 씨. 제닝스 Delivery of biologically active polypeptides
CN1944463A (en) * 2006-10-30 2007-04-11 中国科学技术大学 Fusion protein with alpha-interferon activity and its coded gene and use
CN102007144A (en) * 2006-12-29 2011-04-06 斯洛柏塔盖兹欧洲有限公司 Microvesicles derived from recombinant yeast having haemostatic activities and uses thereof
CN104313048A (en) * 2014-10-21 2015-01-28 长沙中科晶博生物科技有限公司 Method for producing lactoferrin by using saccharomyces cerevisiae
CN107109386A (en) * 2014-10-27 2017-08-29 丹尼斯科美国公司 The composition related to β glucosidases and method

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL71991A (en) * 1983-06-06 1994-05-30 Genentech Inc Preparation of mature human IGF and EGF via prokaryotic recombinant DNA technology
AU4329793A (en) * 1992-06-30 1994-01-24 Viagen Oy (lactobacillus) expression system using surface protein gene sequences
JP5354559B2 (en) * 2005-11-24 2013-11-27 独立行政法人産業技術総合研究所 Highly efficient secretory signal peptides and protein expression systems using them
WO2015199441A1 (en) * 2014-06-24 2015-12-30 한국생명공학연구원 Translational fusion partner for secretory production of target protein derived from pichia pastoris strain and use thereof
US20160222372A1 (en) * 2015-01-30 2016-08-04 The Government Of The United States Of America, As Represented By The Secretary Of The Navy Enzyme/Protein Packaged Bacterial Vesicles for Therapeutic Delivery
KR20160110232A (en) * 2015-03-11 2016-09-21 주식회사 엠디헬스케어 Composition for Prevention or Treatment of Inflammatory disease Comprising Extracellular Vesicles Derived from Lactic acid bacteria
US20160331686A1 (en) * 2015-05-12 2016-11-17 Clsn Laboratories, Inc. Compositions and Methods for Yeast Extracellular Vesicles as Delivery Systems
KR102394638B1 (en) * 2015-09-30 2022-05-09 (주)아모레퍼시픽 Composition comprising lactic acid bacteria derived extracellular vesicles for preventing hair loss or promoting hair growth
KR20180003344A (en) * 2016-06-30 2018-01-09 (주)아모레퍼시픽 Anti-inflammatory composition comprising extracellular vesicles derived from yeast

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5821079A (en) * 1985-05-02 1998-10-13 Transgene S.A. Vectors for the expression and secretion of hirudin by transformed yeasts
KR19990064308A (en) * 1995-10-20 1999-07-26 알. 씨. 제닝스 Delivery of biologically active polypeptides
CN1944463A (en) * 2006-10-30 2007-04-11 中国科学技术大学 Fusion protein with alpha-interferon activity and its coded gene and use
CN102007144A (en) * 2006-12-29 2011-04-06 斯洛柏塔盖兹欧洲有限公司 Microvesicles derived from recombinant yeast having haemostatic activities and uses thereof
CN104313048A (en) * 2014-10-21 2015-01-28 长沙中科晶博生物科技有限公司 Method for producing lactoferrin by using saccharomyces cerevisiae
CN107109386A (en) * 2014-10-27 2017-08-29 丹尼斯科美国公司 The composition related to β glucosidases and method

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Comparison of Alpha-Factor Preprosequence and a Classical Mammalian Signal Peptide for Secretion of Recombinant Xylanase xynB from Yeast Pichia pastoris;He Zuyong等;《J. Microbiol. Biotechnol》;20120212;第22卷(第4期);479-483页 *
Potential of yeast secretory vesicles in biodelivery systems;Gurusamy Kutralam-Muniasamy等;《Drug Discovery Today》;20150630;第20卷(第6期);摘要部分、第663页-664页、表1 *
Species-specific variation in signal peptide design Implications for protein secretion in foreign hosts;Gunnar von Heijne等;《FEBS LETTERS》;19890228;第244卷(第2期);第439-446页 *
张嗣良等.信号肽.《多尺度微生物过程优化》.化学工业出版社,2003, *

Also Published As

Publication number Publication date
CN112074264A (en) 2020-12-11
US20210169802A1 (en) 2021-06-10
KR102015116B1 (en) 2019-10-21
JP2023134441A (en) 2023-09-27
JP7297872B2 (en) 2023-06-26
WO2019212293A1 (en) 2019-11-07
EP3773423A1 (en) 2021-02-17
EP3773423A4 (en) 2022-04-27
JP2021521898A (en) 2021-08-30

Similar Documents

Publication Publication Date Title
CN112074264B (en) Extracellular vesicles derived from recombinant microorganisms comprising polynucleotides encoding target proteins and uses thereof
CN109152737A (en) Nano vesicle of binding matrix and application thereof
WO2009043277A1 (en) Skin care composition containing hsa fusion protein, method for preparation and use thereof
CN102471769A (en) Compositions and methods for genetic modification of cells having cosmetic function to enhance cosmetic appearance
KR102341138B1 (en) Exosomes comprising exosomal membrane protein variant and manufacturing method thereof
US20040043003A1 (en) Clinical grade vectors based on natural microflora for use in delivering therapeutic compositions
JP2024009822A (en) Methods and compositions for treating inflammatory skin disease with recombinant microorganisms
Del Carmen et al. A novel interleukin-10 DNA mucosal delivery system attenuates intestinal inflammation in a mouse model
JP2020023502A (en) Methods and compositions for stimulation of cell proliferation, and provision of biologically active mixtures of fgf2 isoforms
KR100594607B1 (en) Probiotic microorganisms producing human growth hormone fused with Fc fragment of human IgG for oral delivery system and a method for producing the same
US11530414B2 (en) Gene expression cassette and expression vector including the same
KR20170009112A (en) Novel alpha-1,3-glucanse
KR100457879B1 (en) Plasmid originated from Bifidobacterium, recombinant expression vector using the plasmid and transformation method
AU776350B2 (en) Immunostimulant bacterial membrane fractions in cancer treatment
WO2021034131A1 (en) Extracellular vesicles derived from recombinant cells including polynucleotide encoding il-1 inhibitor as target protein and use thereof
JP2000503850A (en) Recombinant expression of S-layer-protein
CN111117967A (en) Method for preparing cell over expressing exogenous gene
US20220111009A1 (en) Exogenous protein-expressing microorganism and use thereof
KR20220000144A (en) Method for isolating subpopulations of exosomes from population of exosomes and its application
WO2022137644A1 (en) Bacterium, il10 gene expression enhancer, immune response control enhancer, and preparation
Demolder et al. Human interferon-β, expressed in Saccharomyces cerevisiae, is predominantly directed to the vacuoles Influence of modified co-expression of secretion factors and chaperones
KR101860040B1 (en) Acidic Fibroblast Growth Factors with Skin Physiological Activity Produced by Plant Cell Culture System
CN117925712A (en) In-vivo protein overexpression method of scylla paramamosain based on bacterial extracellular vesicles
JP2022033444A (en) Germ, il10 gene expression promoting agent, and pharmaceutical preparation
CN110938647A (en) Recombinant expression vector, recombinant attenuated salmonella typhimurium and construction method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant